You are on page 1of 9

PERSPECTIVE

published: 05 July 2019


doi: 10.3389/fgene.2019.00638

Epigenetic Regulation at the


Interplay Between Gut Microbiota
and Host Metabolism
Joan Miro-Blanch 1,2 and Oscar Yanes 1,2*
1 Metabolomics Platform, IISPV, Department of Electronic Engineering, Universitat Rovira i Virgili, Tarragona, Spain,
2 Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain

Gut microbiota communities have coevolved for millions of years in a symbiotic


relationship with their mammalian hosts. Elucidating and understanding the molecular
mechanisms by which microbiota interacts with its host and how this contributes to the
homeostasis of the host is crucial. One of these molecular relationships is the so-called
chemical crosstalk between microbiota and host metabolisms, including the poorly
explored epigenetic regulation of host tissues by the metabolic activity of gut microbiota
in response to changes in diet. DNA methylation and histone modifications are epigenetic
marks partly regulated by enzymes such as methylases and acetylases, whose activity
depend on host and microbiota metabolites that act as substrates and cofactors for
these reactions. However, providing a complete mechanistic description of the regulatory
interactions between both metabolisms and the impact on the expression of host genes
Edited by: through an epigenetic modulation, remains elusive. This article presents our perspective
Jane Mellor, on how metabolomic, metagenomic, transcriptomic, and epigenomic data can be used
University of Oxford, United Kingdom
to investigate the “microbiota–nutrient metabolism–epigenetics axis.” We also discuss the
Reviewed by:
implications and opportunities this knowledge may have for basic and applied science,
Tomas J. Ekström,
Karolinska Institute (KI), Sweden such as the impact on the way we structure future research, understand, and prevent
Thomas Roeder, diseases like type 2 diabetes or obesity.
University of Kiel, Germany

*Correspondence: Keywords: microbiota, epigenetics, metabolomics, metabolism, histones, omics


Oscar Yanes
oscar.yanes@urv.cat
CROSSTALK BETWEEN HOST AND GUT MICROBIOTA
Specialty section: METABOLISMS
This article was submitted to
Epigenomics and Epigenetics, The human body co-habit with a diverse community of symbiotic microorganisms and their set
a section of the journal
of genes, collectively known as the microbiome (Ursell et al., 2013). The acquisition of the initial
Frontiers in Genetics
microbiome is a dynamic rather than a static process during early life (Ferretti et al., 2018). A recent
Received: 18 March 2019 estimation of the number of bacterial cells over human cells in our body has reduced the ratio from
Accepted: 18 June 2019
10:1 (Savage, 1977) to a 1.3:1 (Sender et al., 2016a; Sender et al., 2016b). This implies a similar number
Published: 05 July 2019
of bacterial and human cells in and on the human body. However, the human microbiome encodes
Citation:
for at least 100 times more genes than our genome (Qin et al., 2010). Therefore, the corresponding
Miro-Blanch J and Yanes O (2019)
Epigenetic Regulation at the Interplay
higher functionality of bacterial genes is a key aspect to understand existing metabolic interactions
Between Gut Microbiota and Host between the host and its microbiota.
Metabolism. Front. Genet. 10:638. The microbiota helps their hosts to digest dietary fiber; produces some important neurotransmitters
doi: 10.3389/fgene.2019.00638 (Sampson and Mazmanian, 2015; Yano et al., 2015; Strandwitz et al., 2018), hormones, and vitamins

Frontiers in Genetics  |  www.frontiersin.org 1 July 2019 | Volume 10 | Article 638


Miro-Blanch and Yanes Gut Microbiota, Host Metabolism and Epigenetics

(Kau et al., 2011; Trial et al., 2016); helps in training the host levels of histone acetylation promote decondensation and relaxation
immune system (Kau et al., 2011; Chu and Mazmanian, 2013); of chromatin, supporting a more transcriptionally active state of
and protects against pathogens (Watanabe et al., 2017), among chromatin (Bolduc et al., 2017).
many other functions. However, unbalanced microbiota can also Alteration of chromatin state is a possible mechanism by
cause disease. Some common diseases in western societies such which gut microbiota induces host immune maturation (Cani
as obesity and type 2 diabetes are associated with shifts in the et al., 2012; Seeley et al., 2018). Recognition of a “self ” antigen
relative abundance of gut bacteria composition and functionality, should not only be limited to mammalian host antigens, but
compared to the ones observed in healthy individuals. The cause also symbiotic microbiota antigens forming part of the whole
of the microbiota imbalance (dysbiosis) of unhealthy individuals human ecosystem in a healthy state. The human leukocyte
across age and geography has been mainly correlated with dietary antigen (HLA) or major histocompatibility complex (MHC)
habits (Turnbaugh et al., 2008; De Filippo et al., 2010; Muegge et al., gene system encodes many antigen-presenting proteins, which
2011; Wu et al., 2011; Arumugam et al., 2011; Menni et al., 2017). are essential to recognize and distinguish “self ” from “non-self ”
Therefore, the so-called chemical crosstalk between the microbiota antigens. Colonization of germ-free mice has demonstrated the
and its host has tangible consequences for the physiological state capacity of microbiota-specific species to activate MHC class II
of the host (Sharon et al., 2014; Caesar et al., 2015; Ussar et al., genes (Umesaki et al., 1995). However, little is known about the
2016). However, the molecular mechanisms by which microbiota immunomodulatory effect of microbial metabolites. A poorly
chemically interacts with host cells and regulate gene expression explored possibility is that epigenetically relevant metabolites
remain largely unknown. In this regard, the role that certain host- such as SCFA, highly influenced by microbiota composition,
microbiota derivate metabolites may exert on epigenetic events at would regulate MHC gene expression by coordinating activity of
the DNA, RNA, and histone level needs to be further investigated. enzymes that acetylate and methylate histones and DNA allowing
chromatin accessibility (Ting and Trowsdale, 2002).
Regulation of DNA and histone methylation may be driven
THE “MICROBIOTA–NUTRIENT by complex microbiota–host metabolism interactions involving
METABOLISM–HOST EPIGENETIC” AXIS S-adenosyl methionine (SAM), derived from the essential amino
acid methionine through diet (Poirier et al., 2001). Folate plays
Differences in the microbiota or epigenome in two genetically an essential role by re-methylating homocysteine to methionine
identical organisms, such as same-sex inbred mice or monozygous (Figure 1), thereby ensuring the provision of SAM (Kim, 2005;
twins, can create differences in susceptibilities to diseases Krautkramer et al., 2017). In this regard, enzymes that are depleted in
including  obesity and type 2 diabetes (Ling and Groop, 2009; obese microbiomes are frequently involved in cofactor and vitamin
Franks and Ling, 2010). As with gut microbiota, new studies have metabolism (Greenblum et al., 2012), including the production of
demonstrated that epigenetic events are highly dynamic, changing cobalamin (vitamin B12), pyridoxal phosphate (the active form of
in response to nutrient availability (Bouchard et al., 2010; Hullar vitamin B6), tetrahydrofolate, and folate (Arumugam et al., 2011;
and Fu, 2014) or physical exercise (Laker et al., 2017). DNA Kau et al., 2011; Yatsunenko et al., 2012). Taken together, dysbiosis
methyltransferases, DNA hydroxylases, histone acetyltransferases, of microbiota can influence SAM levels and, as a result, alter the
histone deacetylases, histone methyltransferases, and histone methylation status of DNA and histones. Whether dysbiosis of
demethylases are enzymes responsible for adding or removing microbiota can alter α-ketoglutarate and succinate levels in specific
these dynamic epigenetic modifications. In this regard, peripheral host tissues, and regulate the rate of DNA demethylation,
endogenous metabolites can regulate gene expression through is a plausible but little explored possibility. Ten-eleven translocation
epigenetic events in host cells (Katada et al., 2012). For instance, (TET) enzymes are a key family of DNA and histone demethylases
histone deacetylation regulated by sirtuin family deacetylases that use α-ketoglutarate as co-substrate. However, due to the
is regulated by the NAD+/NADH ratio, acetyl-CoA, O-acetyl- structural similarity with α-ketoglutarate, Tets are susceptible
ADP-ribose, and nicotinamide (Peleg et al., 2016; Ringel et  al., to competitive inhibition by fumarate and succinate, causing an
2018). Whether gut microbiota metabolism is regulating increase in histone and DNA methylation levels (An et al., 2017).
the concentration and/or activity of endogenously produced Another modification that could be regulated by microbial
metabolites by the host remains largely unexplored, and it is only metabolism is histone phosphorylation. In response to a low
recently that an increasing number of researchers have started to ATP/AMP ratio indicative of energy status, the AMP-activated
investigate this possibility (Krautkramer et al., 2016; Aleksandrova protein kinase (AMPK) can translocate to chromatin and
et al., 2017; Romano et al., 2017). Krautkramer and colleagues phosphorylate histone H2B (Bungard et al., 2010). Changes in
(2016) have demonstrated that microbial colonization regulates AMPK activity have been reported in obesity, type 2 diabetes,
global histone acetylation and methylation in multiple host tissues metabolic syndrome, and cardiovascular disease (Kola et al.,
in a diet-dependent manner. 2008). Interestingly, germ-free mice were resistant to obesity
Short-chain fatty acids (SCFAs) are exclusively produced by and insulin resistance that develop after consuming a Western-
the microbial fermentation of dietary carbohydrates, and their style, high-fat, and sugar-rich diet (Bäckhed et al., 2007). The
abundances are regulated by the composition of the microbiota persistently lean phenotype of germ-free animals was associated
(Chen et al., 2007; Cai et al., 2011). Importantly, SCFAs, particularly with increased skeletal muscle and liver levels of phosphorylated
butyrate and acetate, produced by the microbiota, inhibit histone AMPK. It is also tempting to speculate that phosphotransferase
deacetylases (Figure 1) (Maslowski and MacKay, 2011). Increased systems (PTS) overrepresented in the Western diet microbiomes

Frontiers in Genetics  |  www.frontiersin.org 2 July 2019 | Volume 10 | Article 638


Miro-Blanch and Yanes Gut Microbiota, Host Metabolism and Epigenetics

FIGURE 1 | The “microbiota–nutrient metabolism–epigenetics” axis. Most of the key molecules involved in one-carbon metabolism are dietary- and microbiota-
dependent, being susceptible to gut dysbiosis or diet intervention. Folate is the precursor of dihydrofolate (DHF) and tetrahydrofolate (THF), and dietary intake is
the only source for humans. Together with vitamin B12, 5’methy-THF is in charge of remethylating homocysteine (Hcy) to methionine (Met), a crucial step in the
process of transferring a methyl group to DNA or histones through SAM. The ratio of S-adenosyl homocysteine (SAH) to SAM regulates the overall methylation
status of the genome at the DNA or histone level. Vitamins B12, B2, and B6 are key cofactors in the folate cycle that are produced by the microbiota or ingested
through diet. Intermediates of the tricarboxylic acid cycle (TCA) are known to positively or negatively regulate histone methylation. For example, alpha-ketoglutarate
(α-KG) is known to be an essential substrate for jumonji C histone demethylases (jmjC), and levels of succinate and fumarate can inhibit jmjC demethylases.
α-Ketoglutarate is a co-substrate of TET dioxygenases in charge of demethylation processes of histones and DNA. As for jmjC demethylases, increased levels of
fumarate and succinate can inhibit TET enzymes with the consequent increased levels of histone and DNA methylation. Short-chain fatty acids (SCFAs) produced
by the gut microbiota are also known to inhibit or promote histone PTMs. Butyrate and propionate are inhibitors of sirtuins deacetylases enzymes. Acetate from
gut fermentation contributes to the pool of intermediate molecules known to form acetyl-coenzyme A, the major acetyl group donor for histone acetyl transferases
(HATs). Acetate is also known to be an inhibitor of histone deacetylases (HDAC), increasing histone acetylation levels and regulating chromatin accessibility. Whether
levels of FAD/FADH2, NAD/NADH, TCA intermediates, and other host endogenous epigenetically relevant metabolites are modulated by gut microbiota metabolism
needs to be further investigated. DMG, dimethylglycine; ATP, adenosine triphosphate; ADP, adenosine diphosphate; FAD/FADH2, Flavin adenine dinucleotide; NAD+/
NADH, nicotinamide adenine dinucleotide.

(Turnbaugh et al., 2008; Turnbaugh et al., 2009) could have an termed holobiont (Youle et al., 2013; Carrier and Reitzel, 2017;
impact on this histone modification. Groussin et al., 2017; van de Guchte et al., 2018). In this regard,
In short, dysbiosis and reduction of the microbiota diversity we could arguably talk about holo–genome, –transcriptome,
can potentially alter the levels of nutrients and metabolites –proteome, or –metabolome, referring to the combination of
that can potentially act as regulators of DNA methylation and both, host and host microbiota molecular layers or modules
histone modifications either by directly inhibiting enzymes that of information at the DNA, RNA, protein, or metabolite level,
catalyze the processes, or by altering the availability of substrates respectively (Figure 2). To investigate the dynamics of the
necessary for the enzymatic reactions. holobiont ecosystem network, multi-omic approaches bring
unprecedented advantages. Diseases such as obesity and diabetes
are known to be multifactorial, and the collection of several
HOLOBIONTS, MULTIFACTORIAL -omic data (Table 1) from the same holobiont specimen, may
DISEASES, AND OMIC TECHNOLOGIES provide a detailed molecular description and new mechanistic
insights of how dietary nutrients and gut microbiota metabolism
Hosts and their microbiota have a very intimate relationship and can regulate the host phenotype through gene expression and
should be considered as a single biological and evolutionary unit, epigenetic and metabolic regulation.

Frontiers in Genetics  |  www.frontiersin.org 3 July 2019 | Volume 10 | Article 638


Miro-Blanch and Yanes Gut Microbiota, Host Metabolism and Epigenetics

FIGURE 2 | The human holobiont. Representation of few examples of known interactions between different molecular levels within a holobiont. Exercise,
environment, and diet can affect the physiology and molecular interactions between human (host) and its microbiota at the DNA, RNA, protein, or metabolite level.
As an example, fecal host micro RNAs (miRNAs) are used by the host to modulate the composition of its own gut microbiota, interacting at the microbiota RNA and
DNA levels to control microbial growth (Liu et al., 2016). Short-chain fatty acids (SCFAs), products of gut bacterial anaerobic fermentation of dietary fiber, have been
proved to cause changes in histone PTMs in multiple host tissues (Krautkramer et al., 2016). Butyrate is a potent histone deacetylase inhibitor (HDACi), regulating
the transcription levels of genes involved in colorectal tumorigenesis (Hassig et al., 1997). The direct transformation of dietary nutrients (Sharon et al., 2014) and
secondary products of host metabolites such as primary bile acids (Wahlström et al., 2016) evidencies the strong interdependency between host and microbiota.
Folate production by Biffidobacterium spp. is another example of how gut microbiota products can affect epigenetics such as DNA or histone methylation (Paul
et al., 2015). Microbiota diversity shifts, products, or bacterial structural components such as flagellin can cause the activation of the immune system as well as
impact the immune reconstitution after certain diseases or immunotherapy (Manzo and Bhatt, 2015). Immune system maturation and allergic disease development
are other examples of how the host and its microbiome interact (Christmann et al., 2015).

To study a complex metabolic disorder such as familial type mice, regulate histone post-translational modifications (PTMs).
1 diabetes mellitus (T1D), Heintz-Buschart et al. (2016) used Comparing histone PTMs and transcriptional profiles between
a combination of host genomics and proteomics together with conventional, germ-free mice and germ-free mice supplemented
metagenomics, metatranscriptomics, and metaproteomics with SCFAs, Krautkramer et al. concluded that SCFAs alone
to demonstrate a pronounced family membership effect in are partially causative for histone PTMs. In the same direction,
the structuration and functionality of the microbiomes. They Thaiss et al. (2016) used a combination of metagenomics,
observed a correlation between certain human pancreatic transcriptomics, epigenomics, and metabolomics together with
enzymes and the expression of specific microbial genes involved imaging electron microscopy, to identify a diurnal rhythmicity in
in key T1D metabolic transformations. Krautkramer et al. (2016) the microbial biogeography, metabolic profile, and metagenomic
used a combination of metabolomic, proteomics of histones, functionality as critical orchestrators of host epigenetic marks
transcriptomic, and metagenomic techniques in conventional and gene expression. Thaiss et al. have demonstrated that host
and germ-free mice, to demonstrate how SCFAs produced by epigenetic and transcriptional circadian oscillations are partially
the microbiota, or supplemented exogenously to germ-free dependent on environmental signals such as microbiome

Frontiers in Genetics  |  www.frontiersin.org 4 July 2019 | Volume 10 | Article 638


Miro-Blanch and Yanes Gut Microbiota, Host Metabolism and Epigenetics

TABLE 1 | Popular omic techniques in the fields of epigenomics, metagenomics, metabolomics, and proteomics.

Omics Focus Trait studied Techniques used Reference

Epigenomics DNA modifications 5-methylcytosine WGBS (whole genome bisulfite Seq) (Lister et al., 2009)
RRBS (reduced represented bisulfite Seq) (Xi et al., 2012)
MeDIP-Seq (methylated DNA IP Seq) (Down et al., 2008)
5-hydroxymethylation oxBS-Seq (oxidative bisulfite Seq) (Booth et al., 2012)
5-formylcytosine RedBS-Seq (reduced bisulfite Seq) (Booth et al., 2014)
RNA modifications 6-methyladenosine m6A-Seq (m6A specific methylated IP Seq) (Meyer et al., 2012)
DNA 3D structure DNA structure and ChIP-Seq (chromatin IP Seq) (Barski et al., 2007)
protein interaction ATAC-Seq (assay transposase accessible chromatin Seq) (Buenrostro et al., 2013)
Hi-C (chromatin conformation capture) (Lieberman-Aiden et al., 2009)
DNase-Seq (DNase I hypersensitive sites Seq) (Boyle et al., 2008)
RNA transcripts Transcribed DNA RNA-Seq (mRNA/size/strand Seq) (Mortazavi et al., 2008)
GRO-Seq (global run-on-sequencing Seq) (Core et al., 2008)
NET-Seq (native elongating transcript Seq) (Churchman and Weissman, 2011)
UMI method (unique molecular identifiers) (Kivioja et al., 2012)
Metagenomics Marker gene Hypervariable region 16S gene (16S amplicon PCR/sequencing) (Woese and Fox, 1977)
Whole metagenome Whole genome DNA-Seq (regular DNA Seq) (Tyson et al., 2004)
Metatranscriptome RNA RNA-Seq (regular RNA Seq) (Gilbert et al., 2008)
Metabolomics Targeted Known metabolites QqQ (triple quadrupole) (Lu et al., 2008)
Untargeted profiling Unknown metabolites qTOF-MS (quadrupole time of flight) (Patti et al., 2012)
Orbitrap-MS
NMR (nuclear magnetic resonance) (Brindle et al., 2002)
Proteomics Histones PTMs H2A, H2B, H3, and Bottom-up (Sidoli et al., 2012)
H4 modifications Middle-down
Top-down
MALDI-imaging mass spectrometry (Lahiri et al., 2016)

The integration and combination of these techniques have the potential to reveal more mechanistic insights on how gut microbiota influence epigenetics and gene expression,
ultimately affecting host health. IP, immune precipitation; Seq, sequencing; m6A, 6’methyl adenosine; MS, mass spectrometry; LC, liquid chromatography; GC, gas chromatography;
PTMs, posttranslational modifications; MALDI, Matrix-assisted laser desorption ionization.

metabolite dynamics in the intestines and that peripheral organs chemical crosstalk between host and microbiota by uncovering
“sense and adapt” to this circadian metabolite rhythms in a specific epigenetic changes in host cells induced by colonization
similar manner. of specific bacterial strains. Colonizing gnotobiotic mice with
Overall, multi-omic approaches will facilitate the structuration single strains or small bacterial consortia, instead of whole fecal
of future research, improve patient stratification toward a transplants, might bring more accurate information to understand
more personalized care, and open new avenues to evaluate specific molecular mechanisms and molecular pathways involved
the effectiveness of functional probiotics, functional foods, or in the host epigenetic regulations. Microbiota-induced dysbiosis
nutritional interventions aimed at regulating host gene expression with antibiotics might provide a useful approach to validate
in health and disease. findings in gnotobiotic models by partially mimicking the effects
of absence of microbiota or a reduction in the microbial diversity.
Interestingly, organoids might also provide a challenging but
OPPORTUNITIES FOR BIOMEDICAL AND very useful in vitro culturing system to study host–microbiota
CLINICAL RESEARCH interactions (Williamson et al., 2018).
In a clinical context, gaining knowledge on the “microbiota–
The development and improvement of new technologies and nutrient metabolism–host epigenetics axis” using multiple
bioinformatic tools are advancing biomedical research at a fast -omic approaches in combination with microbiota modulation
pace. Multi-omic experiments are allowing researchers to obtain therapies, has the potential to prevent and treat more efficiently
mechanistic insights on the human holobiont homeostasis, metabolic diseases:
improving decision-making for next experiments to be performed.
In a research context, extensive collection of -omic data will allow (1)
Fecal microbiota transplantation (FMT): The use of
integration of information into modulable and controlled models microbiota modulation therapies such as FMT to treat
of microbial communities (Franzosa et al., 2015). However, it is recurrent Clostridium difficile infections has been proved
important to identify confounding factors in longitudinal -omic significantly more efficient than a vancomycin treatment
studies. Standardization of methods and techniques to reduce (Petrof and Khoruts, 2012; van Nood et al., 2013). Recently,
noise and bias in microbiome research will improve how we the potential of FMT as microbiome modulation technique
translate lab findings into the clinic (Knight et al., 2018). for treating metabolic (Kootte et al., 2017) (Gupta et al.,
Using germ-free or gnotobiotic mouse models provide a 2016), neurological (Kang et al., 2017), and immunological
framework to manipulate the gut microbial composition in disorders (Pamer, 2014) has been tested, improving these
a controlled manner. These models can be used to study the conditions by partially restoring microbiota diversity and

Frontiers in Genetics  |  www.frontiersin.org 5 July 2019 | Volume 10 | Article 638


Miro-Blanch and Yanes Gut Microbiota, Host Metabolism and Epigenetics

functionality. Assessing long-term host epigenetic effects of the potential myriad of metabolites that can produce and interact
FMT has to be further investigated. with any cell of our body through systemic circulation. Those
(2) Microbiota as drug target: The unique microbiome metabolites are coming from direct transformations of nutrients
composition of each person is probably responsible for different available in the gut microbiota or from secondary transformed
susceptibilities to the same nutrient, pollutant, or drug treatment host products. The link between epigenetic marks and gut
(Koppel et al., 2017). Microbiota-derived metabolites can enter microbes appears to be mediated by host-microbial metabolites
the bloodstream and interact with drug treatments, impacting that act as substrates and cofactors for key epigenetic enzymes in
the efficacy, toxicity, and clearance of the drug. Diagnosing or the host. A disruption in the composition of the gut microbiota
treating diseases using microbiota-targeted drugs, probiotics, may lead to unbalanced key metabolites that sequentially may
and use of bacteriophages or engineered bacteria has recently impact epigenetic pathways and alter gene expression. The
re-emerged (Bhat and Kapila, 2017; Landry and Tabor, 2017; implementation of multi-omic approaches to study the human
Manrique et al., 2017; Maier et al., 2018). holobiont will facilitate the stratification of patients toward
(3) Nutritional intervention, probiotics, and prebiotics: The acute a personalized-oriented care, improving disease prevention,
consumption of a cocktail of probiotics containing a selection diagnostics, drug election, and treatment efficiency.
of five strains of Lactobacillus and five strains of Enterococcus
modulates the microbiome and enhances SCFAs production
in human and mice (Nagpal et al., 2018). The consumption of DATA AVAILABILITY
some probiotics has proven to be beneficial and ameliorates
stress felt in healthy women (Tillisch Kirsten, 2013), improves All datasets analyzed for this study are cited in the manuscript
insulin sensitivity (Gomes et al., 2014), protects against and/or the supplementary files.
infections (Reid and Burton, 2002), and helps to restore
microbiota after distortion by antibiotics (Langdon et al.,
2016), among other benefits. Promoting SCFAs producing AUTHOR CONTRIBUTIONS
bacteria in the host gut by a nutritional intervention that
JM and OY designed, wrote, revised, and approved the submitted
increases fiber consumption (Zhao et al., 2018) may have an
version of the manuscript.
epigenetic effect in the host (Krautkramer et al., 2016).

FUNDING
CONCLUSIONS AND PERSPECTIVE
This work was supported by the European Union’s Horizon 2020
The study of the “microbiota–nutrient metabolism–host research and innovation program under the Marie Skłodowska-
epigenetic” axis has great potential to reveal the molecular Curie grant agreement No 675610. OY thanks the following
mechanisms by which gut microbiota composition affects the bodies for funding: Ministerio de Economia y Competitividad
expression of genes in their hosts. In a human holobiont context, (MINECO) (BFU2017-87958-P) and the Spanish Biomedical
this axis is relevant to understand, prevent, diagnose, and treat the Research Centre in Diabetes and Associated Metabolic Disorders
existing epidemic of metabolic disorders such as type 2 diabetes (CIBERDEM), an initiative of Instituto de Investigacion
and obesity. Microbiota is a key player in health outcomes due to Carlos III (ISCIII).

REFERENCES Bolduc, J.-F., Hany, L., Barat, C., Ouellet, M., and Tremblaya, M. J. (2017).
Epigenetic metabolite acetate inhibits class I/II histone deacetylases, promotes
Aleksandrova, K., Romero-Mosquera, B., and Hernandez, V. (2017). Diet, gut histone acetylation, and increases HIV-1 Integration in CD4 T cells. J. Virol. 91,
microbiome and epigenetics: emerging links with inflammatory bowel diseases 1–15. doi: 10.1128/JVI.01943-16
and prospects for management and prevention. Nutrients 9, 1–13. doi: 10.3390/ Booth, M. J., Branco, M. R., Ficz, G., Oxley, D., Krueger, F., Reik, W., et al. (2012).
nu9090962 Quantitative sequencing of 5-methylcytosine and 5-hydroxymethylcytosine at
An, J., Rao, A., and Ko, M. (2017). TET family dioxygenases and DNA single-base resolution. Science (80-). 336, 934–937. doi: 10.1126/science.1220671
demethylation in stem cells and cancers. Exp. Mol. Med. 49, 1–12. doi: 10.1038/ Booth, M. J., Marsico, G., Bachman, M., Beraldi, D., and Balasubramanian, S.
emm.2017.5 (2014). Quantitative sequencing of 5-formylcytosine in DNA at single-base
Arumugam, M., Raes, J., Pelletier, E., Le Paslier, D., Yamada, T., Mende, D. R., et al. resolution. Nat. Chem. 6, 435–440. doi: 10.1038/nchem.1893
(2011). Enterotypes of the human gut microbiome. Nature 473, 174–180. doi: Bouchard, L., Rabasa-Lhoret, R., Faraj, M., Lavoie, M. È., Mill, J., Pérusse, L., et al.
10.1038/nature09944 (2010). Differential epigenomic and transcriptomic responses in subcutaneous
Bäckhed, F., Manchester, J. K., Semenkovich, C. F., and Gordon, J. I. (2007). adipose tissue between low and high responders to caloric restriction. Am. J.
Mechanisms underlying the resistance to diet-induced obesity in germ-free Clin. Nutr. 91, 309–320. doi: 10.3945/ajcn.2009.28085
mice. Proc. Natl. Acad. Sci. 104, 979–984. doi: 10.1073/pnas.0605374104 Boyle, A. P., Davis, S., Shulha, H. P., Meltzer, P., Margulies, E. H., Weng, Z., et al.
Barski, A., Wei, G., Chepelev, I., Wang, Z., Cui, K., Zhao, K., et al. (2007). High- (2008). High-resolution mapping and characterization of open chromatin
resolution profiling of histone methylations in the human genome. Cell 129, across the genome. Cell 132, 311–322. doi: 10.1016/j.cell.2007.12.014
823–837. doi: 10.1016/j.cell.2007.05.009 Brindle, J. T., McKilligin, E., Tranter, G., Clarke, S., Holmes, E., Nicholson, J. K.,
Bhat, M. I., and Kapila, R. (2017). Dietary metabolites derived from gut microbiota: et al. (2002). Rapid and noninvasive diagnosis of the presence and severity of
critical modulators of epigenetic changes in mammals. Nutr. Rev. 75, 374–389. coronary heart disease using 1H-NMR-based metabonomics. Nat. Med. 8,
doi: 10.1093/nutrit/nux001 1439–1445. doi: 10.1038/nm802

Frontiers in Genetics  |  www.frontiersin.org 6 July 2019 | Volume 10 | Article 638


Miro-Blanch and Yanes Gut Microbiota, Host Metabolism and Epigenetics

Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y., and Greenleaf, W. J. (2013). Hassig, C. A., Tong, J. K., and Schreiber, S. L. (1997). Fiber-derived butyrate
Transposition of native chromatin for multimodal regulatory analysis and and the prevention of colon cancer. Chem. Biol. 4, 783–789. doi: 10.1016/
personal epigenomics. Nat. Methods 10, 1213–1218. doi: 10.1038/nmeth.2688 S1074-5521(97)90111-3
Bungard, D., Benjamin, J., Fuerth, Zeng, P.-Y., Faubert, B., Maas, N. L., et al. Heintz-Buschart, A., May, P., Laczny, C. C., Lebrun, L. A., Bellora, C., Krishna, A.,
(2010). Signaling kinase AMPK activates stress-promoted transcription via et al. (2016). Integrated multi-omics of the human gut microbiome in a
histone H2B phosphorylation. Science (80-). 329, 1201–1205. doi: 10.1126/ case study of familial type 1 diabetes. Nat. Microbiol. 2, 1–12. doi: 10.1038/
science.1191241 nmicrobiol.2016.180
Caesar, R., Tremaroli, V., Kovatcheva-datchary, P., and Patrice, D. (2015). Crosstalk Hullar, M. A. J., and Fu, B. C. (2014). Diet, the gut microbiome, and epigenetics.
between gut microbiota and dietary lipids aggravates WAT inflammation through Cancer J. (United States) 20, 170–175. doi: 10.1097/PPO.0000000000000053
TLR signaling. Cell Metab., 22, 658–668. doi: 10.1016/j.cmet.2015.07.026 Kang, D. W., Adams, J. B., Gregory, A. C., Borody, T., Chittick, L., Fasano, A.,
Cai, L., Sutter, B. M., Li, B., and Tu, B. P. (2011). Acetyl-CoA induces cell growth et al. (2017). Microbiota transfer therapy alters gut ecosystem and improves
and proliferation by promoting the acetylation of histones at growth genes. gastrointestinal and autism symptoms: an open-label study. Microbiome 5,
Mol. Cell 42, 426–437. doi: 10.1016/j.molcel.2011.05.004 1–16. doi: 10.1186/s40168-016-0225-7
Cani, P. D., Osto, M., Geurts, L., and Everard, A. (2012). Involvement of gut Katada, S., Imhof, A., and Sassone-Corsi, P. (2012). Connecting threads: epigenetics
microbiota in the development of low-grade inflammation and type 2 diabetes and metabolism. Cell 148, 24–28. doi: 10.1016/j.cell.2012.01.001
associated with obesity. Gut Microbes 3, 37–41. doi: 10.4161/gmic.19625 Kau, A. L., Ahern, P. P., Griffin, N. W., Goodman, A. L., and Gordon, J. I. (2011).
Carrier, T. J., and Reitzel, A. M. (2017). The hologenome across environments and Human nutrition, the gut microbiome and the immune system. Nature 474,
the implications of a host-associated microbial repertoire. Front. Microbiol. 8, 327–336. doi: 10.1038/nature10213
1–13. doi: 10.3389/fmicb.2017.00802 Kim, Y.-I. (2005). Nutritional epigenetics: impact of folate deficiency on dna
Chen, Y., Sprung, R., Tang, Y., Ball, H., Sangras, B., Kim, S. C., et al. (2007). Lysine methylation and colon cancer susceptibility. J. Nutr. 135, 2694–2697. doi:
propionylation and butyrylation are novel post-translational modifications in 10.1093/jn/135.11.2703
histones. Mol. Cell. Proteomics 6, 812–819. doi: 10.1074/mcp.M700021-MCP200 Kivioja, T., Vähärautio, A., Karlsson, K., Bonke, M., Enge, M., Linnarsson, S.,
Christmann, B. S., Abrahamsson, T. R., Charles, N., Duck, L. W., Mannon, P. J., et al. (2012). Counting absolute numbers of molecules using unique molecular
Berg, G., et al. (2015). Human seroreactivity to gut microbiota antigens. identifiers. Nat. Methods 9, 72–74. doi: 10.1038/nmeth.1778
J. Allergy Clin. Immunol. 136, 1378–1386. doi: 10.1016/j.jaci.2015.03.036 Knight, R., Navas, J., Quinn, R. A., Sanders, J. G., and Zhu, Q. (2018). Best practices
Chu, H., and Mazmanian, S. K. (2013). Innate immune recognition of the for analysing microbiomes. Nat. Rev. Microbiol. 16, 410–422. doi: 10.1038/
microbiota promotes host-microbial symbiosis. Nat. Immunol. 14, 668–675. s41579-018-0029-9
doi: 10.1038/ni.2635 Kola, B., Grossman, A. B., and Korbonits, M. (2008). The role of AMP-activated
Churchman, L. S., and Weissman, J. S. (2011). Nascent transcript sequencing protein kinase in obesity. Front. Horm. Res. 36, 198–211. doi: 10.1159/000115366
visualizes transcription at nucleotide resolution. Nature 469, 1–18. doi: Kootte, R. S., Levin, E., Salojärvi, J., Smits, L. P., Hartstra, A. V., Udayappan, S. D.,
10.1038/nature09652 et al. (2017). Improvement of insulin sensitivity after lean donor feces in
Core, L. J., Waterfall, J. J., and Lis, J. T. (2008). Nascent RNA sequencing metabolic syndrome is driven by baseline intestinal microbiota composition.
reveals  widespread pausing and divergent initiation at human promoters. Cell Metab. 26, 611–619.e6. doi: 10.1016/j.cmet.2017.09.008
Science (80-). 322, 1845–1848. doi: 10.1126/science.1162228 Koppel, N., Rekdal, V. M., and Balskus, E. P. (2017). Chemical transformation of
De Filippo, C., Cavalieri, D., Di Paola, M., Ramazzotti, M., Poullet, J. B., xenobiotics by the human gut microbiota. Science (80-). 356, 1246–1257. doi:
Massart, S., et al. (2010). Impact of diet in shaping gut microbiota revealed by 10.1126/science.aag2770
a comparative study in children from Europe and rural Africa. Proc. Natl. Acad. Krautkramer, K. A., Kreznar, J. H., Romano, K. A., Vivas, E. I., Barrett-Wilt, G. A.,
Sci. 107, 14691–14696. doi: 10.1073/pnas.1005963107 Rabaglia, M. E., et al. (2016). Diet-microbiota interactions mediate global
Down, T. A., Rakyan, V. K., Turner, D. J., Flicek, P., Li, H., Kulesha, E., et al. (2008). epigenetic programming in multiple host tissues. Mol. Cell 64, 982–992. doi:
A bayesian deconvolution strategy for immunoprecipitation-based DNA 10.1016/j.molcel.2016.10.025
methylome analysis. Nat. Biotechnol. 26, 779–785. doi: 10.1038/nbt1414 Krautkramer, K. A., Rey, F. E., and Denu, J. M. (2017). Chemical signaling between
Ferretti, P., Pasolli, E., Tett, A., Asnicar, F., Gorfer, V., Fedi, S., et al. (2018). gut microbiota and host chromatin: What is your gut really saying? J. Biol.
Mother-to-infant microbial transmission from different body sites shapes Chem. 292, 8582–8593. doi: 10.1074/jbc.R116.761577
the developing infant gut microbiome. Cell Host Microbe 24, 133–145.e5. doi: Lahiri, S., Sun, N., Solis-Mezarino, V., Fedisch, A., Ninkovic, J., Feuchtinger, A.,
10.1016/j.chom.2018.06.005 et al. (2016). In situ detection of histone variants and modifications in mouse
Franks, P. W., and Ling, C. (2010). Epigenetics and obesity: the devil is in the brain using imaging mass spectrometry. Proteomics 16, 437–447. doi: 10.1002/
details. BMC Med. 8, 88. doi: 10.1186/1741-7015-8-88 pmic.201500345
Franzosa, E. A., Hsu, T., Sirota-Madi, A., Shafquat, A., Abu-Ali, G., Morgan, Laker, R. C., Garde, C., Camera, D. M., Smiles, W. J., Zierath, J. R., Hawley, J. A.,
X. C., et al. (2015). Sequencing and beyond: integrating molecular “omics” for et al. (2017). Transcriptomic and epigenetic responses to short-term nutrient-
microbial community profiling. Nat. Rev. Microbiol. 13, 360–372. doi: 10.1038/ exercise stress in humans. Sci. Rep. 7, 1–12. doi: 10.1038/s41598-017-15420-7
nrmicro3451 Landry, B. P., and Tabor, J. J. (2017). Engineering diagnostic and therapeutic gut
Gilbert, J. A., Field, D., Huang, Y., Edwards, R. A., Li, W., Gilna, P., et al. (2008). bacteria. Microbiol. Spectr. 5, 1–22. doi: 10.1128/microbiolspec.BAD-0020-2017
Detection of large numbers of novel sequences in the metatranscriptomes Langdon, A., Crook, N., and Dantas, G. (2016). The effects of antibiotics on
of complex marine microbial communities. PLoS One 3, 277–286. doi: the microbiome throughout development and alternative approaches for
10.1002/9781118010549.ch27 therapeutic modulation. Genome Med. 8, 1–16. doi: 10.1186/s13073-016-0294-z
Gomes, A. C., Bueno, A. A., Souza, R. G. M., and Mota, J. F. (2014). Gut Lieberman-Aiden, E., Berkum5, van, N. L., Williams, L., Imakaev2, M.,
microbiota, probiotics and diabetes. J. Clin. Gastroenterol. 13, 1–13. doi: Ragoczy, T., et al. (2009). Comprehensive mapping of long range interactions
10.1097/MCG.0000000000001058 reveals folding principles of the human genome. Science (80-). 326, 289–293.
Greenblum, S., Turnbaugh, P. J., and Borenstein, E. (2012). Metagenomic systems doi: 10.1126/science.1181369
biology of the human gut microbiome reveals topological shifts associated with Ling, C., and Groop, L. (2009). Epigenetics: a molecular link between environmental
obesity and in flammatory bowel disease. Proc. Natl. Acad. Sci. 109, 594–599. factors and type 2 diabetes. Diabetes 58, 2718–2725. doi: 10.2337/db09-1003
doi: 10.1073/pnas.1116053109 Lister, R., Pelizzola, M., Dowen, R. H., Hawkins, R. D., Hon, G., Nery,
Groussin, M., Mazel, F., Sanders, J. G., Smillie, C. S., Lavergne, S., Thuiller, W., et al. J.  R., et al. (2009). Human DNA methylomes at base resolution show
(2017). Unraveling the processes shaping mammalian gut microbiomes over widespreadepigenomic differences. Nature 462, 315–322. doi: 10.1038/
evolutionary time. Nat. Commun. 8, 1–12. doi: 10.1038/ncomms14319 nature08514
Gupta, S., Allen-Vercoe, E., and Petrof, E. O. (2016). Fecal microbiota transplantation: Liu, S., Da Cunha, A. P., Rezende, R. M., Cialic, R., Wei, Z., Bry, L., et al. (2016).
In perspective. Therap. Adv. Gastroenterol. 9, 229–239. doi: 10.1177/​175​ The host shapes the gut microbiota via fecal MicroRNA. Cell Host Microbe 19,
6283X15607414 32–43. doi: 10.1016/j.chom.2015.12.005

Frontiers in Genetics  |  www.frontiersin.org 7 July 2019 | Volume 10 | Article 638


Miro-Blanch and Yanes Gut Microbiota, Host Metabolism and Epigenetics

Lu, W., Bennett, B. D., and Rabinowitz, J. D. (2008). Analytical strategies for Seeley, J. J., Baker, R., Mohamed, G., Bruns, T., Hayden, M. S., Deshmukh, S. D.,
LC-MS-based targeted metabolomics. J. Chromatogr. B. Analyt. Technol. et al. (2018). Induction of innate immune memory via microRNA targeting
Biomed. Life Sci. 871, 236–242. doi: 10.1016/j.jchromb.2008.04.031 of chromatin remodeling factors. Nature 559, 114–119. doi: 10.1038/
Maier, L., Pruteanu, M., Kuhn, M., Zeller, G., Telzerow, A., Anderson, E. E., et al. s41586-018-0253-5
(2018). Extensive impact of non-antibiotic drugs on human gut bacteria. Sender, R., Fuchs, S., and Milo, R. (2016a). Are we really vastly outnumbered?
Nature 555, 623–628. doi: 10.1038/nature25979 Revisiting the ratio of bacterial to host cells in humans. Cell 164, 337–340. doi:
Manrique, P., Dills, M., and Young, M. J. (2017). The human gut phage community 10.1016/j.cell.2016.01.013
and its implications for health and disease. Viruses 9, 9–11. doi: 10.3390/ Sender, R., Fuchs, S., and Milo, R. (2016b). Revised estimates for the number of
v9060141 human and bacteria cells in the body. PLoS Biol. 14, 1–14. doi: 10.1371/journal.
Manzo, V. E., and Bhatt, A. S. (2015). The human microbiome in hematopoiesis and pbio.1002533
hematologic disorders. Blood 126, 311–318. doi: 10.1182/blood-2015-04-574392 Sharon, G., Garg, N., Debelius, J., Knight, R., Dorrestein, P. C. and Mazmanian,
Maslowski, K. M., and MacKay, C. R. (2011). Diet, gut microbiota and immune S.  K. (2014). Specialized metabolites from the microbiome in health and
responses. Nat. Immunol. 12, 5–9. doi: 10.1038/ni0111-5 disease. Cell Metab. 20, 719–730. doi: 10.1016/j.cmet.2014.10.016
Menni, C., Jackson, M. A., Pallister, T., Steves, C. J., Spector, T. D., and Valdes, A. Sidoli, S., Cheng, L., and Jensen, O. N. (2012). Proteomics in chromatin biology
M. (2017). Gut microbiome diversity and high-fibre intake are related to lower and epigenetics: elucidation of post-translational modifications of histone
long-term weight gain. Int. J. Obes. 41, 1099–1105. doi: 10.1038/ijo.2017.66 proteins by mass spectrometry. J. Proteomics 75, 3419–3433. doi: 10.1016/j.
Meyer, K. D., Saletore, Y., Zumbo, P., Elemento, O., Mason, C. E., and Jaffrey, S. R. jprot.2011.12.029
(2012). Comprehensive Analysis of mRNA methylation reveals enrichment in Strandwitz, P., Kim, K. H., Terekhova, D., Liu, J. K., Sharma, A., Levering, J.,
3′ UTRs and near stop codons. Cell 149, 1635–1646. doi: 10.1016/j.cell.2012.​ et  al. (2018). GABA-modulating bacteria of the human gut microbiota. Nat.
05.003 Microbiol. 4, 396–403. doi: 10.1038/s41564-018-0307-3
Mortazavi, A., Williams, B. A., McCue, K., Schaeffer, L., and Wold, B. (2008). Thaiss, C. A., Levy, M., Korem, T., Dohnalová, L., Shapiro, H., Jaitin, D. A.,
Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat. et  al. (2016). Microbiota diurnal rhythmicity programs host transcriptome
Methods 5, 621–628. doi: 10.1038/nmeth.1226 oscillations. Cell 167, 1495–1510.e12. doi: 10.1016/j.cell.2016.11.003
Muegge, B. D., Kuczynski, J., Knights, D., Clemente, J. C., Fontana, L., Henrissat, Tillisch, K., Labus, J., Kilpatrick, L., Jiang, Z., Stains, J., Ebrat, B., et al. (2013).
B., et al. (2011). Diet drives convergence in gut microbiome functions across Consumption of Fermented Milk Product with Probiotics Modulates Brain
mammalian phylogeny and within humans. Science (80-). 332, 970–974. doi: Activity. Gastroenterology 144, 1394–1401. doi: 10.1053/j.gastro.2013.02.043
10.1126/science.1198719 Ting, J. P. Y., and Trowsdale, J. (2002). Genetic control of MHC class II expression.
Nagpal, R., Wang, S., Ahmadi, S., Hayes, J., Gagliano, J., Subashchandrabose, S., Cell 109, 21–33. doi: 10.1016/S0092-8674(02)00696-7
et al. (2018). Human-origin probiotic cocktail increases short-chain fatty acid Trial, R. D. P., Reijnders, D., Goossens, G. H., Hermes, G. D. A., Zoetendal, E. G.,
production via modulation of mice and human gut microbiome. Sci. Rep. 8, Dejong, C. H. C., et al. (2016). Effects of gut microbiota manipulation by
1–15. doi: 10.1038/s41598-018-30114-4 antibiotics on host metabolism in obese humans. Cell Metab. 24, 63–74. doi:
Pamer, E. G. (2014). Fecal microbiota transplantation: effectiveness, complexities, 10.1016/j.cmet.2016.06.016
and lingering concerns. Mucosal Immunol. 7, 210–214. doi: 10.1038/mi.​ Turnbaugh, P. J., Bäckhed, F., Fulton, L., and Gordon, J. I. (2008). Diet-induced
2013.117 obesity is linked to marked but reversible alterations in the mouse distal gut
Patti, G. J., Yanes, O., and Siuzdak, G. (2012). Metabolomics: the apogee of the microbiome. Cell Host Microbe 3, 213–223. doi: 10.1016/j.chom.2008.02.015
omics trilogy. Nat. Cell Biol. 13, 263–269. doi: 10.1038/nrm3314 Turnbaugh, P. J., Hamady, M., Yatsunenko, T., Cantarel, B. L., Duncan, A., Ley,
Paul, B., Barnes, S., Demark-Wahnefried, W., Morrow, C., Salvador, C., Skibola, C., R. E., et al. (2009). A core gut microbiome in obese and lean twins. Nature 457,
et al. (2015). Influences of diet and the gut microbiome on epigenetic 480–484. doi: 10.1038/nature07540
modulation in cancer and other diseases. Clin. Epigenetics 7, 1–11. doi: Tyson, G. W., Chapman, J., Hugenholtz, P., Allen, E. E., Ram, R. J., Richardson, P. M.,
10.1186/s13148-015-0144-7 et al. (2004). Community structure and metabolism through reconstruction of
Peleg, S., Feller, C., Ladurner, A. G., and Imhof, A. (2016). The metabolic impact microbial genomes from the environment. Nature 428, 37–43. doi: 10.1038/
on histone acetylation and transcription in ageing. Trends Biochem. Sci. 41, nature02340
700–711. doi: 10.1016/j.tibs.2016.05.008 Umesaki, Y., Okada, Y., Matsumoto, S., Imaoka, A., and Setoyama, H. (1995).
Petrof, E. O., and Khoruts, A. (2012). From stool transplants to next-generation Segmented filamentous bacteria are indigenous intestinal bacteria that activate
microbiota therapeutics. Gastroenterology 1, 233–245. doi: 10.1016/j.dcn.2011.​ intraepithelial lymphocytes and induce MHC Class II molecules and fucosyl
01.002 asialo GM1 glycolipids on the small intestinal epithelial cells in the ex-germ-
Poirier, L. A., Wise, C. K., Delongchamp, R. R., and Sinha, R. (2001). Blood free mouse. Microbiol. Immunol. 39, 555–562. doi: 10.1111/j.1348-0421.1995.
determinations of S -adenosylmethionine, S -adenosylhomocysteine, and tb02242.x
homocysteine: correlations with diet. Cancer Epidemiol. Biomarkers Prev. 10, Ursell, L. K., Metcalf, J. L., Parfrey, L. W., and Knight, R. (2013). Definig the human
649–655. microbiome. Nutr. Rev. 70, S38–S44. doi: 10.1111/j.1753-4887.2012.00493.x
Qin, J., Li, R., Raes, J., Arumugam, M., Burgdorf, S., Manichanh, C., et al. (2010). Ussar, S., Fujisaka, S., and Kahn, C. R. (2016). Interactions between host genetics
A human gut microbial gene catalog established by metagenomic sequencing. and gut microbiome in diabetes and metabolic syndrome. Mol. Metab. 5, 795–
Nature 464, 59–65. doi: 10.1038/nature08821 803. doi: 10.1016/j.molmet.2016.07.004
Reid, G., and Burton, J. (2002). Use of Lactobacillus to prevent infection by van de Guchte, M., Blottière, H. M., and Doré, J. (2018). Humans as holobionts:
pathogenic bacteria. Microbes Infect. 4, 319–324. doi: 10.1016/S1286-4579(02)​ implications for prevention and therapy. Microbiome 6, 81. doi: 10.1186/
01544-7 s40168-018-0466-8
Ringel, A. E., Tucker, S. A., and Haigis, M. C. (2018). Chemical and physiological van Nood, E., Vrieze, A., Nieuwdorp, M., Fuentes, S., Zoetendal, E. G., de Vos,
features of mitochondrial acylation. Mol. Cell 72, 610–624. doi: 10.1016/j. W. M., et al. (2013). Duodenal infusion of donor feces for recurrent clostridium
molcel.2018.10.023 difficile. N. Engl. J. Med. 368, 407–415. doi: 10.1056/NEJMoa1205037
Romano, K. A., Martinez-del Campo, A., Kasahara, K., Chittim, C. L., Vivas, E. I., Wahlström, A., Sayin, S. I., Marschall, H. U., and Bäckhed, F. (2016). Intestinal
Amador-Noguez, D., et al. (2017). Metabolic, epigenetic, and transgenerational crosstalk between bile acids and microbiota and its impact on host metabolism.
effects of gut bacterial choline consumption. Cell Host Microbe 22, 279–290. Cell Metab. 24, 41–50. doi: 10.1016/j.cmet.2016.05.005
doi: 10.1016/j.chom.2017.07.021 Watanabe, K., Gilchrist, C. A., Uddin, M. J., Burgess, S. L., Abhyankar, M. M.,
Sampson, T., and Mazmanian, S. (2015). Control of brain development, function, Moonah, S. N., et al. (2017). Microbiome-mediated neutrophil recruitment
and behavior by the microbiome. Cell Host Microbe 17, 565–576. doi: 10.1016/j. via CXCR2 and protection from amebic colitis. PLoS Pathog. 13, 1–20. doi:
chom.2015.04.011 10.1371/journal.ppat.1006513
Savage, D. D. C. (1977). Microbial ecology of the gastrointestinal tract. Annu. Rev. Williamson, I. A., Arnold, J. W., Samsa, L. A., Gaynor, L., DiSalvo, M., Cocchiaro,
Microbiol. 31, 107–133. doi: 10.1146/annurev.mi.31.100177.000543 J.  L., et al. (2018). A high-throughput organoid microinjection platform to

Frontiers in Genetics  |  www.frontiersin.org 8 July 2019 | Volume 10 | Article 638


Miro-Blanch and Yanes Gut Microbiota, Host Metabolism and Epigenetics

study gastrointestinal microbiota and luminal physiology. Cmgh 6, 301–319. Youle, M., Knowlton, N., Rohwer, F., Gordon, J., and Relman, D. A. (2013).
doi: 10.1016/j.jcmgh.2018.05.004 Superorganisms and holobionts. Microbe Mag. 8, 152–153. doi: 10.1128/
Woese, C. R., and Fox, G. E. (1977). Phylogenetic structure of the prokaryotic microbe.8.152.1
domain: the primary kingdoms. Proc. Natl. Acad. Sci. 74, 5088–5090. doi: Zhao, L., Zhang, F., Ding, X., Wu, G., Lam, Y. Y., Shi, Y., et al. (2018). Gut bacteria
10.1073/pnas.74.11.5088 selectively promoted by dietary fibers alleviate type 2 diabetes. Sci. 1156, 1151–
Wu, G. D., Chen, J., Hoffmann, C., Bittinger, K., Chen, Y., Sue, A., et al. (2011). 1156. doi: 10.1126/science.aao5774
Linking long-term dietary patterns with gut microbial enterotypes. Science
(80-). 334, 105–108. doi: 10.1126/science.1208344 Conflict of Interest Statement: The authors declare that the research was
Xi, Y., Bock, C., Müller, F., Sun, D., Meissner, A., and Li, W. (2012). RRBSMAP: conducted in the absence of any commercial or financial relationships that could
a fast, accurate and user-friendly alignment tool for reduced representation be construed as a potential conflict of interest.
bisulfite sequencing. Bioinformatics 28, 430–432. doi: 10.1093/bioinformatics/
btr668 Copyright © 2019 Miro-Blanch and Yanes. This is an open-access article
Yano, J. M., Yu, K., Donaldson, G. P., Shastri, G. G., Ann, P., Ma, L., et al. distributed under the terms of the Creative Commons Attribution License (CC
(2015). Indigenous bacteria from the gut microbiota regulate host serotonin BY). The use, distribution or reproduction in other forums is permitted, provided
biosynthesis. Cell 161, 264–276. doi: 10.1016/j.cell.2015.02.047 the original author(s) and the copyright owner(s) are credited and that the
Yatsunenko, T., Rey, F. E., Manary, M. J., Trehan, I., Dominguez-Bello, M. G., original publication in this journal is cited, in accordance with accepted academic
Contreras, M., et al. (2012). Human gut microbiome viewed across age and practice. No use, distribution or reproduction is permitted which does not comply
geography. Nature 486, 222–227. doi: 10.1038/nature11053 with these terms.

Frontiers in Genetics  |  www.frontiersin.org 9 July 2019 | Volume 10 | Article 638

You might also like